1
|
Vanders RL, Gomez HM, Daly K, Wark PA, Horvat JC, Hansbro PM. Immune checkpoints are suppressed during pregnancy following influenza A virus infection. Am J Physiol Lung Cell Mol Physiol 2024; 327:L890-L904. [PMID: 39254092 DOI: 10.1152/ajplung.00391.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Influenza A virus (IAV) infection is a major health risk during pregnancy. Although vaccination and antiviral agents are widely used and reduce IAV-induced symptoms, they are not sufficient to control IAV infections in pregnancy, especially during pandemics. Respiratory viruses like IAV exploit immune alterations that occur during pregnancy, including the upregulation of immune checkpoint proteins (ICPs) like programmed death ligand-1 (PDL1), programmed cell death receptor 1 (PD1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA4). We hypothesize that blocking expression of PDL1 on innate immune cells will improve maternal immunity following IAV infection. We used murine models of IAV infection during pregnancy with and without treatment with the immune checkpoint inhibitor (ICI), a-PDL1. Pregnant and nonpregnant mice were infected with mouse-adapted IAV (A/PR/8) and assessed at 3 days post infection (3 dpi). Lung cells were analyzed using flow cytometry. Lung mRNA expression of inflammatory and antiviral markers and histology was measured. Protein concentrations of inflammatory and antiviral markers, as well as viral titers were measured from lung bronchiolar lavage fluid (BALF). Lung function was also assessed. Following IAV infection, immune cells from pregnant mice had significant increases in the ICPs, PDL1, PD1, and CTLA4. a-PDL1 treatment effectively suppressed these ICPs and increased the activation marker, CD86. a-PDL1 treatment also reduced lung inflammatory cell infiltration and viral titers, increased antiviral responses, and improved lung function. Overall, IAV infection in pregnancy activates key inhibitory ICPs, leading to worsened disease outcomes. a-PDL1 treatment during IAV infection in pregnancy is an effective method to reduce ICP expression and improve overall immune cell responses.NEW & NOTEWORTHY Influenza infection worsens disease outcomes during pregnancy; however, treatment with anti-PDL1 can restore immune function during pregnancy.
Collapse
Affiliation(s)
- Rebecca L Vanders
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Henry M Gomez
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Katie Daly
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Peter A Wark
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
2
|
Nguyen THO, Rowntree LC, Chua BY, Thwaites RS, Kedzierska K. Defining the balance between optimal immunity and immunopathology in influenza virus infection. Nat Rev Immunol 2024; 24:720-735. [PMID: 38698083 DOI: 10.1038/s41577-024-01029-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/05/2024]
Abstract
Influenza A viruses remain a global threat to human health, with continued pandemic potential. In this Review, we discuss our current understanding of the optimal immune responses that drive recovery from influenza virus infection, highlighting the fine balance between protective immune mechanisms and detrimental immunopathology. We describe the contribution of innate and adaptive immune cells, inflammatory modulators and antibodies to influenza virus-specific immunity, inflammation and immunopathology. We highlight recent human influenza virus challenge studies that advance our understanding of susceptibility to influenza and determinants of symptomatic disease. We also describe studies of influenza virus-specific immunity in high-risk groups following infection and vaccination that inform the design of future vaccines to promote optimal antiviral immunity, particularly in vulnerable populations. Finally, we draw on lessons from the COVID-19 pandemic to refocus our attention to the ever-changing, highly mutable influenza A virus, predicted to cause future global pandemics.
Collapse
Affiliation(s)
- Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Brendon Y Chua
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
3
|
Liu B, Wang Y, Han G, Zhu M. Tolerogenic dendritic cells in radiation-induced lung injury. Front Immunol 2024; 14:1323676. [PMID: 38259434 PMCID: PMC10800505 DOI: 10.3389/fimmu.2023.1323676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Radiation-induced lung injury is a common complication associated with radiotherapy. It is characterized by early-stage radiation pneumonia and subsequent radiation pulmonary fibrosis. However, there is currently a lack of effective therapeutic strategies for radiation-induced lung injury. Recent studies have shown that tolerogenic dendritic cells interact with regulatory T cells and/or regulatory B cells to stimulate the production of immunosuppressive molecules, control inflammation, and prevent overimmunity. This highlights a potential new therapeutic activity of tolerogenic dendritic cells in managing radiation-induced lung injury. In this review, we aim to provide a comprehensive overview of tolerogenic dendritic cells in the context of radiation-induced lung injury, which will be valuable for researchers in this field.
Collapse
Affiliation(s)
| | - Yilong Wang
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | | | - Maoxiang Zhu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
4
|
Vanders RL, Gomez HM, Hsu AC, Daly K, Wark PAB, Horvat JC, Hansbro PM. Inflammatory and antiviral responses to influenza A virus infection are dysregulated in pregnant mice with allergic airway disease. Am J Physiol Lung Cell Mol Physiol 2023; 325:L385-L398. [PMID: 37463835 DOI: 10.1152/ajplung.00232.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/20/2023] Open
Abstract
Influenza A virus (IAV) infections are increased during pregnancy especially with asthma as a comorbidity, leading to asthma exacerbations, secondary bacterial infections, intensive care unit admissions, and mortality. We aimed to define the processes involved in increased susceptibility and severity of IAV infections during pregnancy, especially with asthma. We sensitized mice to house dust mite (HDM), induced pregnancy, and challenged with HDM to induce allergic airway disease (AAD). At midpregnancy, we induced IAV infection. We assessed viral titers, airway inflammation, lung antiviral responses, mucus hypersecretion, and airway hyperresponsiveness (AHR). During early IAV infection, pregnant mice with AAD had increased mRNA expression of the inflammatory markers Il13 and IL17 and reduced mRNA expression of the neutrophil chemoattractant marker Kc. These mice had increased mucous hyperplasia and increased AHR. miR155, miR574, miR223, and miR1187 were also reduced during early infection, as was mRNA expression of the antiviral β-defensins, Bd1, Bd2, and Spd and IFNs, Ifnα, Ifnβ, and Ifnλ. During late infection, Il17 was still increased as was eosinophil infiltration in the lungs. mRNA expression of Kc was reduced, as was neutrophil infiltration and mRNA expression of the antiviral markers Ifnβ, Ifnλ, and Ifnγ and Ip10, Tlr3, Tlr9, Pkr, and Mx1. Mucous hyperplasia was still significantly increased as was AHR. Early phase IAV infection in pregnancy with asthma heightens underlying inflammatory asthmatic phenotype and reduces antiviral responses.NEW & NOTEWORTHY Influenza A virus (IAV) infection during pregnancy with asthma is a major health concern leading to increased morbidity for both mother and baby. Using murine models, we show that IAV infection in pregnancy with allergic airway disease is associated with impaired global antiviral and antimicrobial responses, increased lung inflammation, mucus hypersecretion, and airway hyperresponsiveness (AHR). Targeting specific β-defensins or microRNAs (miRNAs) may prove useful in future treatments for IAV infection during pregnancy.
Collapse
Affiliation(s)
- Rebecca L Vanders
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Henry M Gomez
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Alan C Hsu
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Katie Daly
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
5
|
Creisher PS, Seddu K, Mueller AL, Klein SL. Biological Sex and Pregnancy Affect Influenza Pathogenesis and Vaccination. Curr Top Microbiol Immunol 2023; 441:111-137. [PMID: 37695427 DOI: 10.1007/978-3-031-35139-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Males and females differ in the outcome of influenza A virus (IAV) infections, which depends significantly on age. During seasonal influenza epidemics, young children (< 5 years of age) and aged adults (65+ years of age) are at greatest risk for severe disease, and among these age groups, males tend to suffer a worse outcome from IAV infection than females. Following infection with pandemic strains of IAVs, females of reproductive ages (i.e., 15-49 years of age) experience a worse outcome than their male counterparts. Although females of reproductive ages experience worse outcomes from IAV infection, females typically have greater immune responses to influenza vaccination as compared with males. Among females of reproductive ages, pregnancy is one factor linked to an increased risk of severe outcome of influenza. Small animal models of influenza virus infection and vaccination illustrate that immune responses and repair of damaged tissue following IAV infection also differ between the sexes and impact the outcome of infection. There is growing evidence that sex steroid hormones, including estrogens, progesterone, and testosterone, directly impact immune responses during IAV infection and vaccination. Greater consideration of the combined effects of sex and age as biological variables in epidemiological, clinical, and animal studies of influenza pathogenesis is needed.
Collapse
Affiliation(s)
- Patrick S Creisher
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD, United States
| | - Kumba Seddu
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD, United States
| | - Alice L Mueller
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD, United States
| | - Sabra L Klein
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD, United States.
| |
Collapse
|
6
|
Influenza Virus Infection during Pregnancy as a Trigger of Acute and Chronic Complications. Viruses 2022; 14:v14122729. [PMID: 36560733 PMCID: PMC9786233 DOI: 10.3390/v14122729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Influenza A virus (IAV) infection during pregnancy disrupts maternal and fetal health through biological mechanisms, which are to date poorly characterised. During pregnancy, the viral clearance mechanisms from the lung are sub-optimal and involve hyperactive innate and adaptive immune responses that generate wide-spread inflammation. Pregnancy-related adaptations of the immune and the cardiovascular systems appear to result in delayed recovery post-viral infection, which in turn promotes a prolonged inflammatory phenotype, increasing disease severity, and causing maternal and fetal health problems. This has immediate and long-term consequences for the mother and fetus, with complications including acute cardiopulmonary distress syndrome in the mother that lead to perinatal complications such as intrauterine growth restriction (IUGR), and birth defects; cleft lip, cleft palate, neural tube defects and congenital heart defects. In addition, an increased risk of long-term neurological disorders including schizophrenia in the offspring is reported. In this review we discuss the pathophysiology of IAV infection during pregnancy and its striking similarity to other well-established complications of pregnancy such as preeclampsia. We discuss general features of vascular disease with a focus on vascular inflammation and define the "Vascular Storm" that is triggered by influenza infection during pregnancy, as a pivotal disease mechanism for short and long term cardiovascular complications.
Collapse
|
7
|
Demery-Poulos C, Romero R, Xu Y, Arenas-Hernandez M, Miller D, Tao L, Galaz J, Farias-Jofre M, Bhatti G, Garcia-Flores V, Seyerle M, Tarca AL, Gomez-Lopez N. Pregnancy imparts distinct systemic adaptive immune function. Am J Reprod Immunol 2022; 88:e13606. [PMID: 35989229 PMCID: PMC9648024 DOI: 10.1111/aji.13606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/05/2022] [Accepted: 07/25/2022] [Indexed: 11/28/2022] Open
Abstract
PROBLEM Pregnancy represents a state of systemic immune activation that is primarily driven by alterations in circulating innate immune cells. Recent studies have suggested that cellular adaptive immune components, T cells and B cells, also undergo changes throughout gestation. However, the phenotypes and functions of such adaptive immune cells are poorly understood. Herein, we utilized high-dimensional flow cytometry and functional assays to characterize T-cell and B-cell responses in pregnant and non-pregnant women. METHODS Peripheral blood mononuclear cells from pregnant (n = 20) and non-pregnant (n = 25) women were used for phenotyping of T-cell and B-cell subsets. T-cell proliferation and B-cell activation were assessed by flow cytometry after in vitro stimulation, and lymphocyte cytotoxicity was evaluated by using a cell-based assay. Statistical comparisons were performed with linear mixed-effects models. RESULTS Pregnancy was associated with modestly enhanced basal activation of peripheral CD4+ T cells. Both CD4+ and CD8+ T cells from pregnant women showed increased activation-induced proliferation; yet, a reduced proportion of these cells expressed activation markers compared to non-pregnant women. There were no differences in peripheral lymphocyte cytotoxicity between study groups. A greater proportion of B cells from pregnant women displayed memory-like and activated phenotypes, and such cells exhibited higher activation following stimulation. CONCLUSION Maternal circulating T cells and B cells display distinct responses during pregnancy. The former may reflect the unique capacity of T cells to respond to potential threats without undergoing aberrant activation, thereby preventing systemic inflammatory responses that can lead to adverse perinatal consequences.
Collapse
Affiliation(s)
- Catherine Demery-Poulos
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Centerfor Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Marcia Arenas-Hernandez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Li Tao
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Jose Galaz
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcelo Farias-Jofre
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Megan Seyerle
- Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
8
|
Serum vitamin D levels and COVID-19 during pregnancy: A systematic review and meta-analysis. Clin Nutr ESPEN 2022; 51:120-127. [PMID: 36184196 PMCID: PMC9461277 DOI: 10.1016/j.clnesp.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/19/2022] [Accepted: 09/02/2022] [Indexed: 11/21/2022]
Abstract
Background Serum vitamin D levels are reported to be associated with the risk of incidence and severity of COVID-19 in the general population. During pregnancy, immune system alterations in line with changes in vitamin D metabolism may affect the course of COVID-19. Thus, we aimed to systematically review the association between vitamin D, pregnancy, and COVID-19. Methods A systematic literature search was conducted in PubMed, Scopus, Web of Science, Embase, and Google Scholar until the end of May 2022. Mean differences (MD) with 95% CI were used as desired effect sizes to assess the association of serum vitamin D levels with the risk of incidence and severity of COVID-19 in pregnant women. Results Among 259 records, 7 and 6 studies were included in the systematic review and meta-analysis, respectively. All included studies had acceptable quality. Our results demonstrated an insignificant difference between infected women and non-infected controls (MD = -2.55 ng/ml, 95% CI: −6.85 – 1.74). But serum vitamin D levels in severe/moderate cases compared to mild ones (MD = −2.71 ng/ml, 95% CI: −4.18 to −1.24) are significantly lower. Conclusion Based on the current evidence, serum vitamin D level does not associate with the risk of SARS-CoV-2 infection among pregnant women, but we find a significant association with the severity of the disease. These findings may be helpful in similar conditions and future studies to better understand the complex immune alterations during pregnancy.
Collapse
|
9
|
Bencze D, Fekete T, Pázmándi K. Correlation between Type I Interferon Associated Factors and COVID-19 Severity. Int J Mol Sci 2022; 23:ijms231810968. [PMID: 36142877 PMCID: PMC9506204 DOI: 10.3390/ijms231810968] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022] Open
Abstract
Antiviral type I interferons (IFN) produced in the early phase of viral infections effectively inhibit viral replication, prevent virus-mediated tissue damages and promote innate and adaptive immune responses that are all essential to the successful elimination of viruses. As professional type I IFN producing cells, plasmacytoid dendritic cells (pDC) have the ability to rapidly produce waste amounts of type I IFNs. Therefore, their low frequency, dysfunction or decreased capacity to produce type I IFNs might increase the risk of severe viral infections. In accordance with that, declined pDC numbers and delayed or inadequate type I IFN responses could be observed in patients with severe coronavirus disease (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), as compared to individuals with mild or no symptoms. Thus, besides chronic diseases, all those conditions, which negatively affect the antiviral IFN responses lengthen the list of risk factors for severe COVID-19. In the current review, we would like to briefly discuss the role and dysregulation of pDC/type I IFN axis in COVID-19, and introduce those type I IFN-dependent factors, which account for an increased risk of COVID-19 severity and thus are responsible for the different magnitude of individual immune responses to SARS-CoV-2.
Collapse
Affiliation(s)
- Dóra Bencze
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, H-4032 Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 1 Egyetem Square, H-4032 Debrecen, Hungary
| | - Tünde Fekete
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, H-4032 Debrecen, Hungary
| | - Kitti Pázmándi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, H-4032 Debrecen, Hungary
- Correspondence: ; Tel./Fax: +36-52-417-159
| |
Collapse
|
10
|
Upregulated influenza A viral entry factors and enhanced interferon-alpha response in the nasal epithelium of pregnant rats. Heliyon 2022; 8:e09407. [PMID: 35592667 PMCID: PMC9111991 DOI: 10.1016/j.heliyon.2022.e09407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 03/02/2022] [Accepted: 05/06/2022] [Indexed: 11/22/2022] Open
Abstract
Despite the increased severity of influenza A infection in pregnancy, knowledge about the expression of cell entry factors for influenza A virus (IAV) and the innate immune response in the nasal epithelium, the primary portal of viral entry, is limited. Here, we compared the expression of IAV cell entry factors and the status of the innate immune response in the nasal epithelium of pregnant vs. non-pregnant female rats. IAV cell entry factors — sialic acid [SA] α-2,3- and α-2,6-linked glycans for avian and human IAV, respectively — were detected and quantified with lectin-based immunoblotting and flow cytometry. Baseline frequencies of innate immune cell phenotypes in single cell suspensions of the nasal epithelium were studied with flow cytometry. Subsequently, the magnitude of interferon and cytokine responses was studied with ELISA and cytokine arrays after intranasal resiquimod, a Toll-like receptor 7/8 agonist that mimics IAV infection. We noted substantially increased expression of cell entry factors for both avian and human IAV in the nasal epithelium during pregnancy. Assessment of the innate immune state of the nasal epithelium during pregnancy revealed two previously unreported features: (i) increased presence of tissue-resident plasmacytoid dendritic cells, and (ii) markedly enhanced release of interferon-α but not of the other interferons or cytokines 2 h after intranasal resiquimod. Collectively, our findings challenge the conventional notion of pregnancy-induced immunosuppression as a cause for severe influenza A disease and suggest the need for focused studies on viral tropism during pregnancy to better understand the proximate cause for the observed immunopathology.
Collapse
|
11
|
Cicalini I, Rossi C, Natale L, Cufaro MC, Catitti G, Vespa S, De Bellis D, Iannetti G, Lanuti P, Bucci I, Stuppia L, De Laurenzi V, Pieragostino D. Passive Immunity to SARS-CoV-2 at Birth Induced by Vaccination in the First Trimester of Pregnancy. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182312789. [PMID: 34886515 PMCID: PMC8657259 DOI: 10.3390/ijerph182312789] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022]
Abstract
As is well known, the COVID-19 infection is affecting the whole world, causing a serious health, social and economic crisis. The viral infection can cause a mild or severe illness, depending on how effectively the virus is countered by the immune system. In this context, the position of pregnant women remains rather unknown. The case described here reports the immune response in a woman in good health and in her newborn son, having undergone complete vaccination during the first trimester of her pregnancy. We performed a serological assay, measuring IgG antibodies to SARS-CoV-2, by a fully automated solid phase DELFIA (time-resolved fluorescence) immunoassay in a few drops of blood, collected by a finger-prick and spotted on filter paper. The dried blood spot (DBS) sample we used is the same type of sample routinely used in a newborn screening program test. Such a simple and minimally invasive approach allowed us to monitor both the mother and the newborn soon after birth for their anti-SARS-CoV-2 IgG levels. The serological test on the DBS carried out on both mother and newborn revealed the presence of anti-SARS-CoV-2 IgG antibodies up to 7 months after vaccination in the mother, and already at 48 h of life in the newborn.
Collapse
Affiliation(s)
- Ilaria Cicalini
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (C.R.); (L.N.); (M.C.C.); (G.C.); (S.V.); (D.D.B.); (G.I.); (P.L.); (I.B.); (L.S.); (V.D.L.); (D.P.)
- Department of Innovative Technologies in Medicine and Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Correspondence: ; Tel.: +39-0871-541333
| | - Claudia Rossi
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (C.R.); (L.N.); (M.C.C.); (G.C.); (S.V.); (D.D.B.); (G.I.); (P.L.); (I.B.); (L.S.); (V.D.L.); (D.P.)
- Department of Psychological, Health and Territory Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Luca Natale
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (C.R.); (L.N.); (M.C.C.); (G.C.); (S.V.); (D.D.B.); (G.I.); (P.L.); (I.B.); (L.S.); (V.D.L.); (D.P.)
| | - Maria Concetta Cufaro
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (C.R.); (L.N.); (M.C.C.); (G.C.); (S.V.); (D.D.B.); (G.I.); (P.L.); (I.B.); (L.S.); (V.D.L.); (D.P.)
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giulia Catitti
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (C.R.); (L.N.); (M.C.C.); (G.C.); (S.V.); (D.D.B.); (G.I.); (P.L.); (I.B.); (L.S.); (V.D.L.); (D.P.)
- Department of Medicine and Aging Science, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Simone Vespa
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (C.R.); (L.N.); (M.C.C.); (G.C.); (S.V.); (D.D.B.); (G.I.); (P.L.); (I.B.); (L.S.); (V.D.L.); (D.P.)
- Department of Medicine and Aging Science, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Domenico De Bellis
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (C.R.); (L.N.); (M.C.C.); (G.C.); (S.V.); (D.D.B.); (G.I.); (P.L.); (I.B.); (L.S.); (V.D.L.); (D.P.)
- Department of Innovative Technologies in Medicine and Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giulia Iannetti
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (C.R.); (L.N.); (M.C.C.); (G.C.); (S.V.); (D.D.B.); (G.I.); (P.L.); (I.B.); (L.S.); (V.D.L.); (D.P.)
| | - Paola Lanuti
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (C.R.); (L.N.); (M.C.C.); (G.C.); (S.V.); (D.D.B.); (G.I.); (P.L.); (I.B.); (L.S.); (V.D.L.); (D.P.)
- Department of Medicine and Aging Science, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Ines Bucci
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (C.R.); (L.N.); (M.C.C.); (G.C.); (S.V.); (D.D.B.); (G.I.); (P.L.); (I.B.); (L.S.); (V.D.L.); (D.P.)
- Department of Medicine and Aging Science, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Liborio Stuppia
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (C.R.); (L.N.); (M.C.C.); (G.C.); (S.V.); (D.D.B.); (G.I.); (P.L.); (I.B.); (L.S.); (V.D.L.); (D.P.)
- Department of Psychological, Health and Territory Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Vincenzo De Laurenzi
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (C.R.); (L.N.); (M.C.C.); (G.C.); (S.V.); (D.D.B.); (G.I.); (P.L.); (I.B.); (L.S.); (V.D.L.); (D.P.)
- Department of Innovative Technologies in Medicine and Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Damiana Pieragostino
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (C.R.); (L.N.); (M.C.C.); (G.C.); (S.V.); (D.D.B.); (G.I.); (P.L.); (I.B.); (L.S.); (V.D.L.); (D.P.)
- Department of Innovative Technologies in Medicine and Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
12
|
Rangchaikul P, Venketaraman V. SARS-CoV-2 and the Immune Response in Pregnancy with Delta Variant Considerations. Infect Dis Rep 2021; 13:993-1008. [PMID: 34940401 PMCID: PMC8700906 DOI: 10.3390/idr13040091] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/23/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022] Open
Abstract
As of September 2021, there has been a total of 123,633 confirmed cases of pregnant women with SARS-CoV-2 infection in the US according to the CDC, with maternal death being 2.85 times more likely, pre-eclampsia 1.33 times more likely, preterm birth 1.47 times more likely, still birth 2.84 times more likely, and NICU admission 4.89 times more likely when compared to pregnant women without COVID-19 infection. In our literature review, we have identified eight key changes in the immunological functioning of the pregnant body that may predispose the pregnant patient to both a greater susceptibility to SARS-CoV-2, as well as a more severe disease course. Factors that may impede immune clearance of SARS-CoV-2 include decreased levels of natural killer (NK) cells, Th1 CD4+ T cells, plasmacytoid dendritic cells (pDC), a decreased phagocytic index of neutrophil granulocytes and monocytes, as well as the immunomodulatory properties of progesterone, which is elevated in pregnancy. Factors that may exacerbate SARS-CoV-2 morbidity through hyperinflammatory states include increases in the complement system, which are linked to greater lung injury, as well as increases in TLR-1 and TLR-7, which are known to bind to the virus, leading to increased proinflammatory cytokines such as IL-6 and TNF-α, which are already elevated in normal pregnant physiology. Other considerations include an increase in angiotensin converting enzyme 2 (ACE2) in the maternal circulation, leading to increased viral binding on the host cell, as well as increased IL-6 and decreased regulatory T cells in pre-eclampsia. We also focus on how the Delta variant has had a concerning impact on SARS-CoV-2 cases in pregnancy, with an increased case volume and proportion of ICU admissions among the infected expecting mothers. We propose that the effects of the Delta variant are due to a combination of (1) the Delta variant itself being more transmissible, contagious, and efficient at infecting host cells, (2) initial evidence pointing to the Delta variant causing a significantly greater viral load that accumulates more rapidly in the respiratory system, (3) the pregnancy state being more susceptible to SARS-CoV-2 infection, as discussed in-depth, and (4) the lower rates of vaccination in pregnant women compared to the general population. In the face of continually evolving strains and the relatively low awareness of COVID-19 vaccination for pregnant women, it is imperative that we continue to push for global vaccine equity.
Collapse
Affiliation(s)
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA;
| |
Collapse
|
13
|
Differential immune responses in pregnant patients recovered from COVID-19. Signal Transduct Target Ther 2021; 6:289. [PMID: 34326311 PMCID: PMC8320317 DOI: 10.1038/s41392-021-00703-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/28/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023] Open
Abstract
Pregnant women are generally more susceptible to viral infection. Although the impact of SARS-CoV-2 in pregnancy remains to be determined, evidence indicates that the risk factors for severe COVID-19 are similar in pregnancy to the general population. Here we systemically analyzed the clinical characteristics of pregnant and non-pregnant female COVID-19 patients who were hospitalized during the same period and found that pregnant patients developed marked lymphopenia and higher inflammation evident by higher C-reactive protein and IL-6. To elucidate the pathways that might contribute to immunopathology or protective immunity against COVID-19 during pregnancy, we applied single-cell mRNA sequencing to profile peripheral blood mononuclear cells from four pregnant and six non-pregnant female patients after recovery along with four pregnant and three non-pregnant healthy donors. We found normal clonal expansion of T cells in the pregnant patients, heightened activation and chemotaxis in NK, NKT, and MAIT cells, and differential interferon responses in the monocyte compartment. Our data present a unique feature in both innate and adaptive immune responses in pregnant patients recovered from COVID-19.
Collapse
|
14
|
Trombetta A, Comar M, Tommasini A, Canton M, Campisciano G, Zanotta N, Cason C, Maso G, Risso FM. SARS-CoV-2 Infection and Inflammatory Response in a Twin Pregnancy. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:3075. [PMID: 33802696 PMCID: PMC8002573 DOI: 10.3390/ijerph18063075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/24/2022]
Abstract
There is growing literature about the SARS-CoV-2 pathogenetic effects exerted during pregnancy and whether vertical transmission or premature birth is possible. It is not well known whether changes in the immune system of pregnant women may lead to a marked susceptibility to infectious processes and the risk of adverse maternal and neonatal complications such as preterm birth, spontaneous abortion, hospitalization in an intensive care unit, transmission to the fetus or newborns, and fetal mortality are poorly understood. Along with this ongoing debate, it is not well defined whether, during pregnancy, the role of host susceptibility in producing a specific inflammatory response to SARS-CoV-2 may represent distinctive markers of risk of vertical transmission. Furthermore, SARS-CoV-2 impact on the vaginal microbiome has not yet been described, despite mounting evidence on its possible effect on the gastrointestinal microbiome and its influence on infectious diseases and preterm labor. This report describes the impact of SARS-CoV-2 on a twin pregnancy diagnosed with infection at the third trimester of gestation including tissue infections, inflammatory response, antibody production, cytokine concentration, and vaginal microbiome composition. We identified a pattern of cytokines including IL1-Ra, IL-9 G-CSF, IL-12, and IL-8 differently expressed, already associated with previously infected patients. We detected a similar concentration of almost all the cytokines tested in both twins, suggesting that the SARS-CoV-2-induced cytokine storm is not substantially impaired during the placental passage. The analysis of the vaginal microbiome did not show relevant signs of dysbiosis, similar to other healthy pregnant women and twin healthy pregnancies. The aim of this report was to analyze the immunological response against SARS-CoV-2 infection and virus tissue tropism in a twin pregnancy.
Collapse
Affiliation(s)
- Andrea Trombetta
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Piazzale Europa, 1, 34127 Trieste, Italy; (A.T.); (M.C.); (M.C.); (C.C.)
| | - Manola Comar
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Piazzale Europa, 1, 34127 Trieste, Italy; (A.T.); (M.C.); (M.C.); (C.C.)
- Institute for Maternal and Child Health “IRCCS Burlo Garofolo”, via Dell’istria 65/1, 34124 Trieste, Italy; (G.C.); (N.Z.); (G.M.); (F.M.R.)
| | - Alberto Tommasini
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Piazzale Europa, 1, 34127 Trieste, Italy; (A.T.); (M.C.); (M.C.); (C.C.)
- Institute for Maternal and Child Health “IRCCS Burlo Garofolo”, via Dell’istria 65/1, 34124 Trieste, Italy; (G.C.); (N.Z.); (G.M.); (F.M.R.)
| | - Melania Canton
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Piazzale Europa, 1, 34127 Trieste, Italy; (A.T.); (M.C.); (M.C.); (C.C.)
| | - Giuseppina Campisciano
- Institute for Maternal and Child Health “IRCCS Burlo Garofolo”, via Dell’istria 65/1, 34124 Trieste, Italy; (G.C.); (N.Z.); (G.M.); (F.M.R.)
| | - Nunzia Zanotta
- Institute for Maternal and Child Health “IRCCS Burlo Garofolo”, via Dell’istria 65/1, 34124 Trieste, Italy; (G.C.); (N.Z.); (G.M.); (F.M.R.)
| | - Carolina Cason
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Piazzale Europa, 1, 34127 Trieste, Italy; (A.T.); (M.C.); (M.C.); (C.C.)
| | - Gianpaolo Maso
- Institute for Maternal and Child Health “IRCCS Burlo Garofolo”, via Dell’istria 65/1, 34124 Trieste, Italy; (G.C.); (N.Z.); (G.M.); (F.M.R.)
| | - Francesco Maria Risso
- Institute for Maternal and Child Health “IRCCS Burlo Garofolo”, via Dell’istria 65/1, 34124 Trieste, Italy; (G.C.); (N.Z.); (G.M.); (F.M.R.)
| |
Collapse
|
15
|
Wastnedge EAN, Reynolds RM, van Boeckel SR, Stock SJ, Denison FC, Maybin JA, Critchley HOD. Pregnancy and COVID-19. Physiol Rev 2021; 101:303-318. [PMID: 32969772 PMCID: PMC7686875 DOI: 10.1152/physrev.00024.2020] [Citation(s) in RCA: 337] [Impact Index Per Article: 84.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/24/2020] [Accepted: 09/22/2020] [Indexed: 01/08/2023] Open
Abstract
There are many unknowns for pregnant women during the coronavirus disease 2019 (COVID-19) pandemic. Clinical experience of pregnancies complicated with infection by other coronaviruses e.g., Severe Acute Respiratory Syndrome (SARS) and Middle Eastern Respiratory Syndrome, has led to pregnant woman being considered potentially vulnerable to severe SARS-CoV-2 infection. Physiological changes during pregnancy have a significant impact on the immune system, respiratory system, cardiovascular function, and coagulation. These may have positive or negative effects on COVID-19 disease progression. The impact of SARS-CoV-2 in pregnancy remains to be determined, and a concerted, global effort is required to determine the effects on implantation, fetal growth and development, labor, and neonatal health. Asymptomatic infection presents a further challenge regarding service provision, prevention, and management. Besides the direct impacts of the disease, a plethora of indirect consequences of the pandemic adversely affect maternal health, including reduced access to reproductive health services, increased mental health strain, and increased socioeconomic deprivation. In this review, we explore the current knowledge of COVID-19 in pregnancy and highlight areas for further research to minimize its impact for women and their children.
Collapse
Affiliation(s)
- Elizabeth A N Wastnedge
- Tommy's Centre for Maternal Health, Medical Research Council (MRC)Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Usher Institute, University of Edinburgh, Edinburgh, United Kingdom; MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Rebecca M Reynolds
- Tommy's Centre for Maternal Health, Medical Research Council (MRC)Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Usher Institute, University of Edinburgh, Edinburgh, United Kingdom; MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Sara R van Boeckel
- Tommy's Centre for Maternal Health, Medical Research Council (MRC)Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Usher Institute, University of Edinburgh, Edinburgh, United Kingdom; MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Sarah J Stock
- Tommy's Centre for Maternal Health, Medical Research Council (MRC)Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Usher Institute, University of Edinburgh, Edinburgh, United Kingdom; MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Fiona C Denison
- Tommy's Centre for Maternal Health, Medical Research Council (MRC)Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Usher Institute, University of Edinburgh, Edinburgh, United Kingdom; MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jacqueline A Maybin
- Tommy's Centre for Maternal Health, Medical Research Council (MRC)Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Usher Institute, University of Edinburgh, Edinburgh, United Kingdom; MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Hilary O D Critchley
- Tommy's Centre for Maternal Health, Medical Research Council (MRC)Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Usher Institute, University of Edinburgh, Edinburgh, United Kingdom; MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
16
|
Abstract
This chapter describes the diagnosis of COVID-19 infection in the general population with special consideration to diagnosis in pregnant women. Diagnosis includes the clinical characteristics including symptoms and signs of infection, similarities and differences between severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and other viral infections particularly influenza, and diagnostic investigations including nucleic acid amplification test, SARS-CoV-2 virus antigen detection, and antibodies against the virus testing. WHO recommendations for testing were discussed. The value of different laboratory investigations in diagnosis and prognosis was highlighted. Explanation of data related to chest imaging and discussion of indications of imaging and different findings were assessed.
Collapse
|
17
|
Honce R, Wohlgemuth N, Meliopoulos VA, Short KR, Schultz-Cherry S. Influenza in High-Risk Hosts-Lessons Learned from Animal Models. Cold Spring Harb Perspect Med 2020; 10:a038604. [PMID: 31871227 PMCID: PMC7706577 DOI: 10.1101/cshperspect.a038604] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Factoring significantly into the global burden of influenza disease are high-risk populations that suffer the bulk of infections. Classically, the very young, very old, and pregnant women have been identified as high-risk populations; however, recent research has uncovered several other conditions that contribute to severe infection. By using varied animal models, researchers have identified molecular mechanisms underpinning the increased likelihood for infection due to obesity and malnourishment, as well as insight into the role sex hormones play in antiviral immunity in males, in females, and across the life span. Additionally, novel comorbidity models have helped elucidate the role of chronic infectious and genetic diseases in influenza virus pathogenesis. Animal models play a vital role in understanding the contribution of host factors to influenza severity and immunity. An in-depth understanding of these host factors represents an important step in reducing the burden of influenza among the growing number of people living with one or more chronic medical conditions.
Collapse
Affiliation(s)
- Rebekah Honce
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105-3678, USA
- Integrated Program in Biomedical Sciences, Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Nicholas Wohlgemuth
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105-3678, USA
| | - Victoria A Meliopoulos
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105-3678, USA
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105-3678, USA
| |
Collapse
|
18
|
Swieboda D, Littauer EQ, Beaver JT, Mills LK, Bricker KM, Esser ES, Antao OQ, Williams DT, Skountzou I. Pregnancy Downregulates Plasmablast Metabolic Gene Expression Following Influenza Without Altering Long-Term Antibody Function. Front Immunol 2020; 11:1785. [PMID: 32922392 PMCID: PMC7457062 DOI: 10.3389/fimmu.2020.01785] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/03/2020] [Indexed: 12/16/2022] Open
Abstract
While the majority of influenza-infected individuals show no or mild symptomatology, pregnant women are at higher risk of complications and infection-associated mortality. Although enhanced lung pathology and dysregulated hormones are thought to underlie adverse pregnancy outcomes following influenza infection, how pregnancy confounds long-term maternal anti-influenza immunity remains to be elucidated. Previously, we linked seasonal influenza infection to clinical observations of adverse pregnancy outcomes, enhanced lung and placental histopathology, and reduced control of viral replication in lungs of infected pregnant mothers. Here, we expand on this work and demonstrate that lower infectious doses of the pandemic A/California/07/2009 influenza virus generated adverse gestational outcomes similar to higher doses of seasonal viruses. Mice infected during pregnancy demonstrated lower hemagglutination inhibition and neutralizing antibody titers than non-pregnant animals until 63 days post infection. These differences in humoral immunity suggest that pregnancy impacts antibody maturation mechanisms without alterations to B cell frequency or antibody secretion. This is further supported by transcriptional analysis of plasmablasts, which demonstrate downregulated B cell metabolism and post-translational modification systems only among pregnant animals. In sum, these findings corroborate a link between adverse pregnancy outcomes and severe pathology observed during pandemic influenza infection. Furthermore, our data propose that pregnancy directly confounds humoral responses following influenza infection which resolves post-partem. Additional studies are required to specify the involvement of plasmablast metabolism with early humoral immunity abnormalities to best guide vaccination strategies and improve our understanding of the immunological consequences of pregnancy.
Collapse
Affiliation(s)
- Dominika Swieboda
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Elizabeth Q Littauer
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Jacob T Beaver
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Lisa K Mills
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Katherine M Bricker
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - E Stein Esser
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Olivia Q Antao
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Dahnide T Williams
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Ioanna Skountzou
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
19
|
Saeed Z, Greer O, Shah NM. Is the Host Viral Response and the Immunogenicity of Vaccines Altered in Pregnancy? Antibodies (Basel) 2020; 9:E38. [PMID: 32759839 PMCID: PMC7551810 DOI: 10.3390/antib9030038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/06/2020] [Accepted: 07/21/2020] [Indexed: 12/11/2022] Open
Abstract
The intricacy of the maternal immune system arises from its ability to prevent a maternal immune response against a semi-allogenic fetus, while protecting the mother against harmful pathogens. However, these immunological adaptations may also make pregnant women vulnerable to developing adverse complications from respiratory viral infections. While the influenza and SARS pandemics support this theory, there is less certainty regarding the clinical impact of SARS-CoV-2 in pregnancy. In the current COVID-19 pandemic, vaccine development is key to public preventative strategies. Whilst most viral vaccines are able to induce a seroprotective antibody response, in some high-risk individuals this may not correlate with clinical protection. Some studies have shown that factors such as age, gender, and chronic illnesses can reduce their effectiveness and in this review, we discuss how pregnancy may affect the efficacy and immunogenicity of vaccines. We present literature to support the hypothesis that pregnant women are more susceptible to respiratory viral infections and may not respond to vaccines as effectively. In particular, we focus on the clinical implications of important respiratory viral infections such as influenza during pregnancy, and the pregnancy induced alterations in important leukocytes such as TFH, cTFH and B cells, which play an important role in generating long-lasting and high-affinity antibodies. Finally, we review how this may affect the efficacy of vaccines against influenza in pregnancy and highlight areas that require further research.
Collapse
Affiliation(s)
| | | | - Nishel Mohan Shah
- Academic Department of Obstetrics & Gynaecology, Imperial College London, Level 3, Chelsea & Westminster Hospital, 369 Fulham Road, London SW10 9NH, UK; (Z.S.); (O.G.)
| |
Collapse
|
20
|
Vazquez-Pagan A, Honce R, Schultz-Cherry S. Impact of influenza virus during pregnancy: from disease severity to vaccine efficacy. Future Virol 2020. [DOI: 10.2217/fvl-2020-0024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Pregnant women are among the individuals at the highest risk for severe influenza virus infection. Infection of the mother during pregnancy increases the probability of adverse fetal outcomes such as small for gestational age, preterm birth and fetal death. Animal models of syngeneic and allogeneic mating can recapitulate the increased disease severity observed in pregnant women and are used to define the mechanism(s) of that increased severity. This review focuses on influenza A virus pathogenesis, the unique immunological landscape during pregnancy, the impact of maternal influenza virus infection on the fetus and the immune responses at the maternal–fetal interface. Finally, we summarize the importance of immunization and antiviral treatment in this population and highlight issues that warrant further investigation.
Collapse
Affiliation(s)
- Ana Vazquez-Pagan
- Graduate School of Biomedical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Rebekah Honce
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, TN, USA
- Integrated Program in Biomedical Sciences, Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
21
|
Brightling CE. Asthma exacerbations during pregnancy: A need for precision medicine. Respirology 2019; 25:670-671. [PMID: 31840898 DOI: 10.1111/resp.13761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 12/05/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Christopher E Brightling
- Institute for Lung Health, Department of Respiratory and Microbial Sciences, Leicester NIHR Biomedical Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
22
|
Vanders RL, Hsu A, Gibson PG, Murphy VE, Wark PAB. Nasal epithelial cells to assess in vitro immune responses to respiratory virus infection in pregnant women with asthma. Respir Res 2019; 20:259. [PMID: 31747925 PMCID: PMC6865028 DOI: 10.1186/s12931-019-1225-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/29/2019] [Indexed: 01/01/2023] Open
Abstract
Respiratory virus-induced asthma exacerbations occur frequently during pregnancy and are associated with adverse outcomes for mother and child. Primary nasal epithelial cells (pNECs) provide a useful method to study immune responses in pregnancy. pNECs were obtained by nasal brushings from pregnant and non-pregnant women with and without asthma. pNECS were infected in vitro with major group Rhinovirus 43 (RV43) and seasonal influenza (H3N2). Following infection, pNECs showed measurable quantities of interferon (IFN)-λ, IL-1β, IL-8, IP-10 and MIP1-α. pNECs provide a safe and effective method for studying respiratory epithelial cell responses during pregnancy.
Collapse
Affiliation(s)
- Rebecca L Vanders
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia. .,Viruses, Infection & Immunity, Vaccines & Asthma (VIVA), Hunter Medical Research Institute, Newcastle, NSW, Australia.
| | - Alan Hsu
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia.,Viruses, Infection & Immunity, Vaccines & Asthma (VIVA), Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Peter G Gibson
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia.,The Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Vanessa E Murphy
- Viruses, Infection & Immunity, Vaccines & Asthma (VIVA), Hunter Medical Research Institute, Newcastle, NSW, Australia.,Priority Research Centre GrowUpWell™, The University of Newcastle, Callaghan, NSW, 2305, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia.,The Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| |
Collapse
|
23
|
Hong S, Banchereau R, Maslow BSL, Guerra MM, Cardenas J, Baisch J, Branch DW, Porter TF, Sawitzke A, Laskin CA, Buyon JP, Merrill J, Sammaritano LR, Petri M, Gatewood E, Cepika AM, Ohouo M, Obermoser G, Anguiano E, Kim TW, Nulsen J, Nehar-Belaid D, Blankenship D, Turner J, Banchereau J, Salmon JE, Pascual V. Longitudinal profiling of human blood transcriptome in healthy and lupus pregnancy. J Exp Med 2019; 216:1154-1169. [PMID: 30962246 PMCID: PMC6504211 DOI: 10.1084/jem.20190185] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/28/2019] [Accepted: 03/04/2019] [Indexed: 12/22/2022] Open
Abstract
Healthy and uncomplicated lupus pregnancies exhibit early and sustained transcriptional modulation of lupus-related pathways. This might contribute to fetal tolerance while predisposing pregnant women to certain infections. Failure to modulate these pathways is associated with lupus pregnancy complications. Systemic lupus erythematosus carries an increased risk of pregnancy complications, including preeclampsia and fetal adverse outcomes. To identify the underlying molecular mechanisms, we longitudinally profiled the blood transcriptome of 92 lupus patients and 43 healthy women during pregnancy and postpartum and performed multicolor flow cytometry in a subset of them. We also profiled 25 healthy women undergoing assisted reproductive technology to monitor transcriptional changes around embryo implantation. Sustained down-regulation of multiple immune signatures, including interferon and plasma cells, was observed during healthy pregnancy. These changes appeared early after embryo implantation and were mirrored in uncomplicated lupus pregnancies. Patients with preeclampsia displayed early up-regulation of neutrophil signatures that correlated with expansion of immature neutrophils. Lupus pregnancies with fetal complications carried the highest interferon and plasma cell signatures as well as activated CD4+ T cell counts. Thus, blood immunomonitoring reveals that both healthy and uncomplicated lupus pregnancies exhibit early and sustained transcriptional modulation of lupus-related signatures, and a lack thereof associates with adverse outcomes.
Collapse
Affiliation(s)
- Seunghee Hong
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY.,Department of Pediatrics, Weill Cornell Medicine, New York, NY.,Baylor Institute for Immunology Research, Dallas, TX
| | - Romain Banchereau
- Baylor Institute for Immunology Research, Dallas, TX.,Oncology Biomarker Development, Genentech, South San Francisco, CA
| | | | - Marta M Guerra
- Department of Medicine and Program in Inflammation and Autoimmunity, Hospital for Special Surgery, New York, NY
| | | | - Jeanine Baisch
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY.,Department of Pediatrics, Weill Cornell Medicine, New York, NY.,Baylor Institute for Immunology Research, Dallas, TX
| | - D Ware Branch
- University of Utah Health Sciences Center, Salt Lake City, UT.,Intermountain Healthcare, Salt Lake City, UT
| | - T Flint Porter
- University of Utah Health Sciences Center, Salt Lake City, UT.,Intermountain Healthcare, Salt Lake City, UT
| | - Allen Sawitzke
- University of Utah Health Sciences Center, Salt Lake City, UT
| | - Carl A Laskin
- Mount Sinai Hospital and the University of Toronto, Toronto, Ontario, Canada
| | - Jill P Buyon
- New York University School of Medicine, New York, NY
| | - Joan Merrill
- Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Lisa R Sammaritano
- Department of Medicine and Program in Inflammation and Autoimmunity, Hospital for Special Surgery, New York, NY.,Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Michelle Petri
- Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | - Marina Ohouo
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY.,Department of Pediatrics, Weill Cornell Medicine, New York, NY.,Baylor Institute for Immunology Research, Dallas, TX
| | | | | | - Tae Whan Kim
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY.,Department of Pediatrics, Weill Cornell Medicine, New York, NY.,Baylor Institute for Immunology Research, Dallas, TX
| | - John Nulsen
- University of Connecticut School of Medicine, Farmington, CT
| | | | | | - Jacob Turner
- Baylor Institute for Immunology Research, Dallas, TX
| | | | - Jane E Salmon
- Department of Medicine and Program in Inflammation and Autoimmunity, Hospital for Special Surgery, New York, NY.,Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Virginia Pascual
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY.,Department of Pediatrics, Weill Cornell Medicine, New York, NY.,Baylor Institute for Immunology Research, Dallas, TX
| |
Collapse
|
24
|
Vangeti S, Yu M, Smed-Sörensen A. Respiratory Mononuclear Phagocytes in Human Influenza A Virus Infection: Their Role in Immune Protection and As Targets of the Virus. Front Immunol 2018; 9:1521. [PMID: 30018617 PMCID: PMC6037688 DOI: 10.3389/fimmu.2018.01521] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022] Open
Abstract
Emerging viruses have become increasingly important with recurrent epidemics. Influenza A virus (IAV), a respiratory virus displaying continuous re-emergence, contributes significantly to global morbidity and mortality, especially in young children, immunocompromised, and elderly people. IAV infection is typically confined to the airways and the virus replicates in respiratory epithelial cells but can also infect resident immune cells. Clearance of infection requires virus-specific adaptive immune responses that depend on early and efficient innate immune responses against IAV. Mononuclear phagocytes (MNPs), comprising monocytes, dendritic cells, and macrophages, have common but also unique features. In addition to being professional antigen-presenting cells, MNPs mediate leukocyte recruitment, sense and phagocytose pathogens, regulate inflammation, and shape immune responses. The immune protection mediated by MNPs can be compromised during IAV infection when the cells are also targeted by the virus, leading to impaired cytokine responses and altered interactions with other immune cells. Furthermore, it is becoming increasingly clear that immune cells differ depending on their anatomical location and that it is important to study them where they are expected to exert their function. Defining tissue-resident MNP distribution, phenotype, and function during acute and convalescent human IAV infection can offer valuable insights into understanding how MNPs maintain the fine balance required to protect against infections that the cells are themselves susceptible to. In this review, we delineate the role of MNPs in the human respiratory tract during IAV infection both in mediating immune protection and as targets of the virus.
Collapse
Affiliation(s)
- Sindhu Vangeti
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Meng Yu
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
25
|
Vermillion MS, Nelson A, Vom Steeg L, Loube J, Mitzner W, Klein SL. Pregnancy preserves pulmonary function following influenza virus infection in C57BL/6 mice. Am J Physiol Lung Cell Mol Physiol 2018; 315:L517-L525. [PMID: 29847990 DOI: 10.1152/ajplung.00066.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Pregnancy is associated with significant anatomic and functional changes to the cardiopulmonary system. Using pregnant C57BL/6 mice, we characterized changes in pulmonary structure and function during pregnancy in healthy animals and following infection with influenza A virus (IAV). We hypothesized that pregnancy-associated alterations in pulmonary physiology would contribute to the more severe outcome of IAV infection. Nonpregnant and pregnant females (at embryonic day 10.5) were either mock-infected or infected with 2009 H1N1 IAV for assessment of pulmonary function, structure, and inflammation at 8 days postinoculation. There were baseline differences in pulmonary function, with pregnant females having greater lung compliance, total lung capacity, and fixed lung volume than nonpregnant females. Following IAV infection, both pregnant and nonpregnant females exhibited reduced circulating progesterone, which in nonpregnant females was associated with increased pulmonary resistance and decreased lung compliance, minute ventilation, and oxygen diffusing capacity compared with uninfected nonpregnant females. In pregnant females, reduced concentrations of progesterone were associated with adverse pregnancy outcomes, but measures of pulmonary function were preserved following IAV infection and were not significantly different from uninfected pregnant mice. Following IAV infection, infectious virus titers and total numbers of pulmonary leukocytes were similar between pregnant and nonpregnant females, but the histological density of pulmonary inflammation was reduced in pregnant animals. These data suggest that pregnancy in mice is associated with significant alterations in pulmonary physiology but that these changes served to preserve lung function during IAV infection. Pregnancy-associated alterations in pulmonary physiology may serve to protect females during severe influenza.
Collapse
Affiliation(s)
- Meghan S Vermillion
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland.,Department of Molecular and Comparative Pathobiology, The Johns Hopkins School of Medicine , Baltimore, Maryland
| | - Andrew Nelson
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland
| | - Landon Vom Steeg
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland
| | - Jeffery Loube
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland
| | - Wayne Mitzner
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland.,Department of Biochemistry and Molecular Biology, The Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland
| |
Collapse
|
26
|
Li J, Lu J, Zhang S, Wang J, Wang H, Liu F, Fang M, Duan X, Liu W. Differential immune response of influenza A virus-infected dendritic cells and association with autophagy. Future Virol 2017. [DOI: 10.2217/fvl-2017-0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: We sought to study the responses of dendritic cells (DCs) after direct stimulation by different influenza A viruses. Materials & methods: Using bone marrow-derived DCs (BMDCs) as a model, we measured the expression of surface markers, cytokine production and the priming effect on CD4+ naive T cells. Results & conclusion: We found that all of the tested viruses induced BMDC maturation. Cytokine expression assays also demonstrated that activated BMDCs secrete higher levels of cytokines. Similar to the maturation degree, well-stimulated BMDCs induced higher levels of naive CD4+ T-cell activation. Furthermore, we found that the PR8 and WSN influenza A viruse-induced BMDC functional activation was at least partially influenced by autophagy.
Collapse
Affiliation(s)
- Jing Li
- CAS Key Laboratory of Pathogenic Microbiology & Immunology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Jiao Lu
- CAS Key Laboratory of Pathogenic Microbiology & Immunology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Shuang Zhang
- CAS Key Laboratory of Pathogenic Microbiology & Immunology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Jing Wang
- CAS Key Laboratory of Pathogenic Microbiology & Immunology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Haoyu Wang
- CAS Key Laboratory of Pathogenic Microbiology & Immunology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
- School of Life Sciences, Anhui University, Hefei 230601, China
| | - Fei Liu
- CAS Key Laboratory of Pathogenic Microbiology & Immunology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Min Fang
- CAS Key Laboratory of Pathogenic Microbiology & Immunology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Xuefeng Duan
- CAS Key Laboratory of Pathogenic Microbiology & Immunology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology & Immunology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| |
Collapse
|
27
|
van Riel D, Mittrücker HW, Engels G, Klingel K, Markert UR, Gabriel G. Influenza pathogenicity during pregnancy in women and animal models. Semin Immunopathol 2016; 38:719-726. [PMID: 27387428 PMCID: PMC7101682 DOI: 10.1007/s00281-016-0580-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 07/01/2016] [Indexed: 11/30/2022]
Abstract
Pregnant women are at the highest risk to develop severe and even fatal influenza. The high vulnerability of women against influenza A virus infections during pregnancy was repeatedly highlighted during influenza pandemics including the pandemic of this century. In 2009, mortality rates were particularly high among otherwise healthy pregnant women. However, our current understanding of the molecular mechanisms involved in severe disease development during pregnancy is still very limited. In this review, we summarize the knowledge on the clinical observations in influenza A virus-infected pregnant women. In addition, knowledge obtained from few existing experimental infections in pregnant animal models is discussed. Since clinical data do not provide in-depth information on the pathogenesis of severe influenza during pregnancy, adequate animal models are urgently required that mimic clinical findings. Studies in pregnant animal models will allow the dissection of involved molecular disease pathways that are key to improve patient management and care.
Collapse
Affiliation(s)
- Debby van Riel
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
- Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Geraldine Engels
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
- Department of Obstetrics and Fetal Medicine, Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Karin Klingel
- Department of Molecular Pathology, University Hospital Tübingen, Tübingen, Germany
| | - Udo R Markert
- Department of Obstetrics and Gynecology, University Hospital Jena, Jena, Germany
| | - Gülsah Gabriel
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany.
- University of Lübeck, Lübeck, Germany.
| |
Collapse
|
28
|
Vanders RL, Murphy VE. Maternal complications and the management of asthma in pregnancy. ACTA ACUST UNITED AC 2015; 11:183-91. [PMID: 25776292 DOI: 10.2217/whe.14.69] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Pregnancy is a unique state requiring alterations in maternal physiology to accommodate the growing fetus. Whilst the maternal immune system is normally well adept at performing this task, the presence of immune disorders, such as asthma, often lead to pregnancy-related complications affecting both mother and baby. Australia has a high prevalence of asthma; with approximately 12% of pregnant women reported to have current asthma. Poor control of asthma is of far greater risk than the use of asthma medications. Being able to identify complications associated with asthma during pregnancy is of great importance in providing appropriate asthma management and medical care to these pregnant women, which may have lifelong consequences for their offspring.
Collapse
Affiliation(s)
- Rebecca L Vanders
- Centre for Asthma & Respiratory Diseases, University of Newcastle & Hunter Medical Research Institute, Newcastle, NSW, Australia
| | | |
Collapse
|
29
|
Kay AW, Blish CA. Immunogenicity and Clinical Efficacy of Influenza Vaccination in Pregnancy. Front Immunol 2015; 6:289. [PMID: 26089824 PMCID: PMC4455389 DOI: 10.3389/fimmu.2015.00289] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 05/19/2015] [Indexed: 11/13/2022] Open
Abstract
Pregnant women are at high risk from influenza due to disproportionate morbidity, mortality, and adverse pregnancy outcomes following infection. As such, they are classified as a high-priority group for vaccination. However, changes in the maternal immune system required to accommodate the allogeneic fetus may alter the immunogenicity of influenza vaccines. A large number of studies have evaluated the safety of the influenza vaccine. Here, we will review available studies on the immunogenicity and efficacy of the influenza vaccine during pregnancy, focusing on both humoral and cellular immunity.
Collapse
Affiliation(s)
- Alexander W Kay
- Blish Laboratory, Department of Pediatrics, Stanford University School of Medicine , Stanford, CA , USA
| | - Catherine A Blish
- Blish Laboratory, Department of Medicine and Stanford Immunology, Stanford University School of Medicine , Stanford, CA , USA
| |
Collapse
|
30
|
The placental protein syncytin-1 impairs antiviral responses and exaggerates inflammatory responses to influenza. PLoS One 2015; 10:e0118629. [PMID: 25831059 PMCID: PMC4382184 DOI: 10.1371/journal.pone.0118629] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 01/21/2015] [Indexed: 12/22/2022] Open
Abstract
Background Pregnancy increases susceptibility to influenza. The placenta releases an immunosuppressive endogenous retroviral protein syncytin-1. We hypothesised that exposure of peripheral monocytes (PBMCs) to syncytin-1 would impair responses to H1N1pdm09 influenza. Methods and Findings Recombinant syncytin-1 was produced. PBMCs from non-pregnant women (n=10) were exposed to H1N1pdm09 in the presence and absence of syncytin-1 and compared to responses of PBMCs from pregnant women (n=12). PBMCs were characterised using flow cytometry, release of interferon (IFN)-α, IFN-λ, IFN-γ, IL-10, IL-2, IL-6 and IL-1β were measured by cytometric bead array or ELISA. Exposure of PBMCs to H1N1pdm09 resulted in the release of IFN-α, (14,787 pg/mL, 95% CI 7311-22,264 pg/mL) IFN-λ (1486 pg/mL, 95% CI 756-2216 pg/mL) and IFN-γ (852 pg/mL, 95% CI 193-1511 pg/mL) after 48 hours. This was significantly impaired in pregnant women (IFN-α; p<0.0001 and IFN-λ; p<0.001). Furthermore, in the presence of syncytin-1, PBMCs demonstrated marked reductions in IFN-α and IFN-λ, while enhanced release of IL-10 as well as IL-6 and IL-1β. Conclusions Our data indicates that a placental derived protein, syncytin-1 may be responsible for the heightened vulnerability of pregnant women to influenza.
Collapse
|
31
|
Vanders RL, Murphy VE, Gibson PG, Hansbro PM, Wark PAB. CD8 T cells and dendritic cells: key players in the attenuated maternal immune response to influenza infection. J Reprod Immunol 2014; 107:1-9. [PMID: 25453203 DOI: 10.1016/j.jri.2014.09.051] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 08/01/2014] [Accepted: 09/09/2014] [Indexed: 12/16/2022]
Abstract
Pregnancy provides a unique challenge for maternal immunity, requiring the ability to tolerate the presence of a semi-allogeneic foetus, and yet still being capable of inducing an immune response against invading pathogens. To achieve this, numerous changes must occur in the activity and function of maternal immune cells throughout the course of pregnancy. Respiratory viruses take advantage of these changes, altering the sensitive balance of maternal immunity, leaving the mother with increased susceptibility to viral infections and increased disease severity. Influenza virus is one of the most common respiratory virus infections during pregnancy, leading to an increased risk of ICU hospitalisations, pneumonia, acute respiratory distress syndrome and even death. Whilst much research has been performed to understand the changes that must take place in maternal immunity during pregnancy, considerable work is still needed to fully comprehend this tremendous feat. To date, few studies have focused on the alterations that occur in maternal immunity during respiratory virus infections. This review highlights the role of dendritic cells (DCs) and CD8 T cells during pregnancy, and the changes that occur in these antiviral cells following influenza virus infections.
Collapse
Affiliation(s)
- Rebecca L Vanders
- Centre for Asthma and Respiratory Diseases, The University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia.
| | - Vanessa E Murphy
- Centre for Asthma and Respiratory Diseases, The University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia.
| | - Peter G Gibson
- Centre for Asthma and Respiratory Diseases, The University of Newcastle, Newcastle, NSW, Australia; The Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia.
| | - Philip M Hansbro
- Centre for Asthma and Respiratory Diseases, The University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia.
| | - Peter A B Wark
- Centre for Asthma and Respiratory Diseases, The University of Newcastle, Newcastle, NSW, Australia; The Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia.
| |
Collapse
|
32
|
Enhanced natural killer-cell and T-cell responses to influenza A virus during pregnancy. Proc Natl Acad Sci U S A 2014; 111:14506-11. [PMID: 25246558 DOI: 10.1073/pnas.1416569111] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pregnant women experience increased morbidity and mortality after influenza infection, for reasons that are not understood. Although some data suggest that natural killer (NK)- and T-cell responses are suppressed during pregnancy, influenza-specific responses have not been previously evaluated. Thus, we analyzed the responses of women that were pregnant (n = 21) versus those that were not (n = 29) immediately before inactivated influenza vaccination (IIV), 7 d after vaccination, and 6 wk postpartum. Expression of CD107a (a marker of cytolysis) and production of IFN-γ and macrophage inflammatory protein (MIP) 1β were assessed by flow cytometry. Pregnant women had a significantly increased percentage of NK cells producing a MIP-1β response to pH1N1 virus compared with nonpregnant women pre-IIV [median, 6.66 vs. 0.90% (P = 0.0149)] and 7 d post-IIV [median, 11.23 vs. 2.81% (P = 0.004)], indicating a heightened chemokine response in pregnant women that was further enhanced by the vaccination. Pregnant women also exhibited significantly increased T-cell production of MIP-1β and polyfunctionality in NK and T cells to pH1N1 virus pre- and post-IIV. NK- and T-cell polyfunctionality was also enhanced in pregnant women in response to the H3N2 viral strain. In contrast, pregnant women had significantly reduced NK- and T-cell responses to phorbol 12-myristate 13-acetate and ionomycin. This type of stimulation led to the conclusion that NK- and T-cell responses during pregnancy are suppressed, but clearly this conclusion is not correct relative to the more biologically relevant assays described here. Robust cellular immune responses to influenza during pregnancy could drive pulmonary inflammation, explaining increased morbidity and mortality.
Collapse
|
33
|
Abstract
Influenza is a major health problem worldwide. Both seasonal influenza and pandemics take a major toll on the health and economy of our country. The present review focuses on the virology and complex immunology of this RNA virus in general and in relation to pregnancy. The goal is to attempt to explain the increased morbidity and mortality seen in infection during pregnancy. We discuss elements of innate and adaptive immunity as well as placental cellular responses to infection. In addition, we delineate findings in animal models as well as human disease. Increased knowledge of maternal and fetal immunologic responses to influenza is needed. However, enhanced understanding of nonimmune, pregnancy-specific factors influencing direct interaction of the virus with host cells is also important for the development of more effective prevention and treatment options in the future.
Collapse
MESH Headings
- Adaptive Immunity
- Animals
- Disease Models, Animal
- Female
- Host-Pathogen Interactions
- Humans
- Immune System/immunology
- Immune System/virology
- Immunity, Innate
- Immunization
- Influenza Vaccines/therapeutic use
- Influenza, Human/immunology
- Influenza, Human/mortality
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Orthomyxoviridae/immunology
- Orthomyxoviridae/pathogenicity
- Pregnancy
- Pregnancy Complications, Infectious/immunology
- Pregnancy Complications, Infectious/mortality
- Pregnancy Complications, Infectious/prevention & control
- Pregnancy Complications, Infectious/virology
- Prognosis
- Risk Factors
Collapse
Affiliation(s)
- Renju S Raj
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine, Burlington, VT, USA
| | - Elizabeth A Bonney
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine, Burlington, VT, USA
| | - Mark Phillippe
- Department of Obstetrics & Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|